WO2004058175A2 - Nouvelle utilisation d'agonistes des recepteurs hepatiques x - Google Patents

Nouvelle utilisation d'agonistes des recepteurs hepatiques x Download PDF

Info

Publication number
WO2004058175A2
WO2004058175A2 PCT/US2003/040906 US0340906W WO2004058175A2 WO 2004058175 A2 WO2004058175 A2 WO 2004058175A2 US 0340906 W US0340906 W US 0340906W WO 2004058175 A2 WO2004058175 A2 WO 2004058175A2
Authority
WO
WIPO (PCT)
Prior art keywords
lxr
subject
agonist
cell
methods
Prior art date
Application number
PCT/US2003/040906
Other languages
English (en)
Other versions
WO2004058175A3 (fr
Inventor
Enrique Saez
Peter Tontonoz
Bryan A. Laffitte
Jing Li
Original Assignee
Irm Llc
The Regents Of The University Of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Irm Llc, The Regents Of The University Of California filed Critical Irm Llc
Priority to AU2003301216A priority Critical patent/AU2003301216A1/en
Publication of WO2004058175A2 publication Critical patent/WO2004058175A2/fr
Publication of WO2004058175A3 publication Critical patent/WO2004058175A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6897Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids involving reporter genes operably linked to promoters
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds

Definitions

  • the present invention generally relates to methods for modulating expression of glucose metabolism-related genes and to methods for treating diabetes mellitus and related disorders. More particularly, the invention pertains to use of LXR agonists to modulate expression of glut4 and other glucose metabolism-related genes, and to enhance glucose uptake and/or reduce gluconeogenesis in subjects suffering from type II diabetes.
  • Type II or noninsulin-dependent diabetes mellitus is a polygenic disease and accounts for >90% of diabetes cases. This disease is characterized by resistance to insulin action on glucose uptake and impaired insulin action to inhibit hepatic glucose production.
  • LXRs Liver X receptors
  • LXR proteins Two LXR proteins ( ⁇ and ⁇ ) are known to exist in mammals. The expression of LXR ⁇ is restricted, with the highest levels being found in the liver, and lower levels found in kidney, intestine, spleen, and adrenals (Willy et al., Genes Dev. 9: 1033-45, 1995). LXR ⁇ is rather ubiquitous, being found in nearly all tissues examined. LXR ⁇ and LXR ⁇ are closely related and share 77% amino acid identity in both their DNA- and ligand-binding domains. The LXRs are also conserved between humans and other animals (e.g., rodents).
  • LXRs heterodimerize with retinoid X receptor (RXR) for function.
  • LXRs are known to be activated by certain naturally occurring, oxidized derivatives of cholesterol, including 22(R)-hydroxycholesterol, 24(S)- hydroxycholesterol and 24,25(S)-epoxycholesterol (see Lehmann et al., J. Biol. Chem. 272: 3137-3140, 1997).
  • the present invention provides methods for enhancing glut4 expression in a cell.
  • the methods entail (i) providing a cell expressing glut4 gene, and (ii) administering to the cell an LXR agonist.
  • Some of the methods are directed to enhancing glut4 expression in adipose cells.
  • the LXR agonist employed is an LXR ⁇ agonist, e.g., GW3965, F3MethylAA, or T0901317.
  • Some of the methods can further comprise measuring glut4 expression level in the cell before and/or after administering the LXR agonist.
  • the cell is present in a subject (e.g., a mammal).
  • the subject is suffering from type II diabetes.
  • the subject can be administered with a pharmaceutical composition comprising an effective amount of the LXR agonist.
  • the subject is administered simultaneously with a known anti-diabetic drug to the subject, e.g., metformin.
  • the invention provides methods for enhancing glut4 expression level in a cell.
  • the methods comprise (i) screening test agents to identify an LXR agonist, and (ii) administering to the cell an effective amount of the LXR agonist; thereby enhancing glut4 expression level in the cell.
  • the cell is an adipose cell.
  • the cell is present in a subject (e.g., a mammal).
  • the subject is suffering from type II diabetes.
  • the LXR agonist is an LXR ⁇ agonist.
  • the present invention provides methods for ameliorating type II diabetes in a subject. These methods entail (i) screening test agents to identify an LXR agonist, and (ii) administering to the subject an effective amount of the LXR agonist; thereby ameliorating type II diabetes in the subject. Additionally, the methods can include measuring circulating glucose level in the subject before and/or after administering the LXR agonist. In some of these methods, the LXR agonist employed is an LXR ⁇ agonist. In some methods, the LXR agonist is administered to the subject at least daily for at least 14 days. Some of the methods can further comprise administering to the subject a known anti-diabetic drug.
  • the invention provides methods for enhancing insulin sensitivity and glucose uptake by a cell in a subject.
  • the methods comprise administering to the subject an effective amount of an LXR agonist; thereby enhancing insulin sensitivity and glucose uptake by the cell.
  • the subject is suffering from type II diabetes.
  • the LXR agonist employed is an LXR ⁇ agonist, e.g., GW3965.
  • the invention provides methods for reducing gluconeogenesis in a subject.
  • the methods entail (i) screening test agents to identify an LXR agonist, and (ii) administering to the subject an effective amount of the LXR agonist; thereby reducing gluconeogenesis in the subject.
  • Some of the methods are directed to subjects who are suffering from type II diabetes.
  • the LXR agonist employed is an LXR ⁇ agonist, e.g., GW3965.
  • the LXR agonist employed in these methods can enhance expression of glucokinase gene, or inhibit expression of at least one of several other gluconeogenesis-related genes (e.g., PGC-1, PEPCK, or glucose-6- ⁇ hosphatase) in liver of the subject.
  • gluconeogenesis-related genes e.g., PGC-1, PEPCK, or glucose-6- ⁇ hosphatase
  • Figures 1A-1B show coordinated regulation of genes involved in glucose metabolism by LXR agonist in liver and white adipose tissue.
  • Figures 2A-2B show activity of LXR ligands on gene expression in skeletal muscle and white adipose.
  • Figures 3A-3B show that LXR agonist effects on expression of glucose metabolism-related genes are dependent on LXR expression.
  • FIG. 4 shows that fasting does not alter expression of LXRs.
  • Figures 5A-5B show that LXR agonists regulate PGC-1 and GLUT4 expression in a cell autonomous manner.
  • Figures 6A-6B show modulation of Glut4 expression in macrophages by LXR ligands.
  • Figure 7 shows sequence alignment of LXREs in the mouse and human GLUT4 promoters (SEQ ID NOs: 1 and 2).
  • Figures 8 A-8B show that the GLUT4 promoter is a direct target for regulation by LXR/RXR heterodimers.
  • Figure 9 shows that an LXR ligand promotes glucose uptake in 3T3-L1 adipocytes.
  • Figures 10A-10B show that an LXR ligand improves glucose tolerance in a model of diet-induced obesity and insulin resistance.
  • Figure 11 shows a synergistic effect between an LXR ligand and a known anti-diabetic drug in reducing circulating glucose level.
  • the present invention is predicated in part on the unexpected discovery that LXR agonists improve glucose tolerance and enhance glut4 expression.
  • the present inventors discovered that there is a coordinate regulation of genes involved in glucose metabolism in liver and adipose tissue. In the liver, LXR agonists inhibit expression of several genes that are important for hepatic gluconeogenesis, e.g., PGC-1, phosphoenolpyruvate carboxykinase (PEPCK), and glucose-6-phosphatase expression.
  • PGC-1 phosphoenolpyruvate carboxykinase
  • PEPCK phosphoenolpyruvate carboxykinase
  • the present invention provides methods for enhancing glut4 expression in cells in a subject by administering an LXR agonist to the subject.
  • the LXR agonist can be any of the LXR agonists known in the art.
  • novel LXR agonists can be screened for administering to the subject.
  • the present invention also provides methods for treating diabetes mellitus and related disorders, such as obesity or hyperglycemia, by administering to a subject an effective amount of an LXR agonist to ameliorate the symptoms of the disease.
  • type II diabetes is amenable to treatment with methods of the present invention.
  • administration with an LXR agonist can also treat other diseases characterized by insulin dysfunction (e.g., resistance, inactivity or deficiency) and/or insufficient glucose transport into cells.
  • the present inventors found that LXR agonists regulate expression levels of a number of genes that play important roles in liver gluconeogenesis. Accordingly, the present invention further provides methods for reducing gluconeogenesis in a subject by modulating expression of such genes (e.g., PGC-1 and PEPCK). These methods comprise (i) screening test agents to identify an LXR agonist and (ii) administering to the subject an effective amount of the LXR agonist. The methods may further comprise detecting a modulatory effect of the LXR agonist on expression of one of these genes in a liver cell of the subject.
  • agent includes any substance, molecule, element, compound, entity, or a combination thereof. It includes, but is not limited to, e.g., protein, polypeptide, small organic molecule, polysaccharide, polynucleotide, and the like. It can be a natural product, a synthetic compound, or a chemical compound, or a combination of two or more substances. Unless otherwise specified, the terms “agent”, “substance”, and “compound” can be used interchangeably.
  • analog is used herein to refer to a molecule that structurally resembles a reference molecule but which has been modified in a targeted and controlled manner, by replacing a specific substituent of the reference molecule with an alternate substituent. Compared to the reference molecule, an analog would be expected, by one skilled in the art, to exhibit the same, similar, or improved utility. Synthesis and screening of analogs, to identify variants of known compounds having improved traits (such as higher binding affinity for a target molecule) is an approach that is well known in pharmaceutical chemistry.
  • contacting has its normal meaning and refers to combining two or more agents (e.g., polypeptides or small molecule compounds) or combining agents and cells. Contacting can occur in vitro, e.g., combining two or more agents or combining a test agent and a cell or a cell lysate in a test tube or other container. Contacting can also occur in a cell or in situ, e.g., contacting two polypeptides in a cell by coexpression in the cell of recombinant polynucleo tides encoding the two polypeptides, or in a cell lysate.
  • agents e.g., polypeptides or small molecule compounds
  • heterologous sequence or a “heterologous nucleic acid,” as used herein, is one that originates from a source foreign to the particular host cell, or, if from the same source, is modified from its original form.
  • a heterologous gene in a host cell includes a gene that, although being endogenous to the particular host cell, has been modified. Modification of the heterologous sequence can occur, e.g., by treating the DNA with a restriction enzyme to generate a DNA fragment that is capable of being operably linked to the promoter. Techniques such as site-directed mutagenesis are also useful for modifying a heterologous nucleic acid.
  • homologous when referring to proteins and/or protein sequences indicates that they are derived, naturally or artificially, from a common ancestral protein or protein sequence.
  • nucleic acids and/or nucleic acid sequences are homologous when they are derived, naturally or artificially, from a common ancestral nucleic acid or nucleic acid sequence. Homology is generally inferred from sequence similarity between two or more nucleic acids or proteins (or sequences thereof). The precise percentage of similarity between sequences that is useful in establishing homology varies with the nucleic acid and protein at issue, but as little as 25% sequence similarity is routinely used to establish homology.
  • sequence similarity percentages e.g., BLASTP and BLASTN using default parameters
  • a "host cell,” as used herein, refers to a prokaryotic or eukaryotic cell into which a heterologous polynucleotide can be or has been introduced.
  • the heterologous polynucleotide can be introduced into the cell by any means, e.g., electroporation, calcium phosphate precipitation, microinjection, transformation, viral infection, and/or the like.
  • sequence identical in the context of two nucleic acid sequences or amino acid sequences refers to the residues in the two sequences which are the same when aligned for maximum correspondence over a specified comparison window.
  • nucleic acid or amino acid sequence means that a nucleic acid or amino acid sequence comprises a sequence that has at least 90% sequence identity or more, preferably at least 95%, more preferably at least 98% and most preferably at least 99%, compared to a reference sequence using any of the programs described in the art (preferably BLAST) using standard parameters.
  • the BLASTP program uses as defaults a wordlength (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff & Henikoff, Proc. Natl. Acad. Sci. USA 89:10915 (1989)). Percentage of sequence identity is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
  • the substantial identity exists over a region of the sequences that is at least about 50 residues in length, more preferably over a region of at least about 100 residues, and most preferably the sequences are substantially identical over at least about 150 residues. In a most preferred embodiment, the sequences are substantially identical over the entire length of the coding regions.
  • LXR liver X receptor
  • LXR receptor includes all subtypes of this receptor. Specifically LXR includes LXR ⁇ and LXR ⁇ . LXR ⁇ has been referred to under a variety of names such as LXRU, LXRa, LXR, RLD-1, NR1H3. It encompasses any polypeptide encoded by a gene with substantial sequence identity to GenBank accession number U22662. Similarly, LXR ⁇ included any polypeptide encoded by a gene referred to as LXRb, LXRP, LXR ⁇ , NER, NERl, UR, OR-1, RIP 15, NR1H2 or a gene with substantial sequence identity to GenBank accession number U07132.
  • ligand refers to an agonist or partial agonist of LXR.
  • the ligand may be selective for LXR ⁇ or LXR ⁇ , or it may have mixed binding affinity for both LXR ⁇ and LXR ⁇ . While a ligand can either agonize or antagonize a receptor function, unless otherwise specified, an LXR ligand used herein primarily refers to an LXR agonist that activated the LXR receptor activities.
  • nucleic acid or “polynucleotide” refers to a deoxyribonucleotide or ribonucleotide polymer in either single- or double-stranded form, and unless otherwise limited, encompasses known analogues of natural nucleotides that hybridize to nucleic acids in manner similar to naturally occurring nucleotides.
  • modulate with respect to an LXR receptor refers to activation of the LXR receptor and its biological activities associated with the LXR pathway (e.g., transcription regulation of a target gene). Modulation of LXR receptor can be up-regulation (i.e., agonizing, activation or stimulation) or down-regulation (i.e. antagonizing, inhibition or suppression).
  • the mode of action of an LXR modulator can be direct, e.g., through binding to the LXR receptor as a ligand.
  • the modulation can also be indirect, e.g., through binding to and/or modifying another molecule which otherwise binds to and activates the LXR receptor.
  • modulation of LXR includes a change in the bioactivities of an LXR agonist ligand (i.e., its activity in binding to and/or activating an LXR receptor) or a change in the cellular level of the ligand.
  • oligonucleotide refers to an oligomer or polymer of ribonucleic acid or deoxyribonucleic acid. This term includes oligonucleotides composed of naturally- occurring nucleobases, sugars and covalent intersugar (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced binding to target and increased stability in the presence of nucleases.
  • operably linked refers to a functional relationship between two or more polynucleotide (e.g., DNA) segments. Typically, it refers to the functional relationship of a transcriptional regulatory sequence to a transcribed sequence.
  • a promoter or enhancer sequence is operably linked to a coding sequence if it stimulates or modulates the transcription of the coding sequence in an appropriate host cell or other expression system.
  • promoter transcriptional regulatory sequences that are operably linked to a transcribed sequence are physically contiguous to the transcribed sequence, i.e., they are cis-acting.
  • a polylinker provides a convenient location for inserting coding sequences so the genes are operably linked to a promoter.
  • Polylinkers are polynucleotide sequences that comprise a series of three or more closely spaced restriction endonuclease recognition sequences.
  • polypeptide is used interchangeably herein with the terms “polypeptides” and “protein(s)”, and refers to a polymer of amino acid residues, e.g., as typically found in proteins in nature.
  • a “mature protein” is a protein which is full-length and which, optionally, includes glycosylation or other modifications typical for the protein in a given cell membrane.
  • screening for LXR agonists refers to use of an appropriate assay system to identify novel LXR agonists from test agents.
  • the assay can be an in vitro or an in vivo assay suitable for identifying whether a test agent can stimulate or activate one or more of the biological functions of the LXR receptor.
  • suitable bioassays include, but are not limited to, assays for examining binding of test agents to an LXR polypeptide (e.g., LXR fragment containing its ligand binding domain), transcription- based assays, creatine kinase assays, assays based on the differentiation of pre-adipocytes, assays based on glucose uptake control in adipocytes, and immunological assays.
  • LXR polypeptide e.g., LXR fragment containing its ligand binding domain
  • transcription- based assays e.g., LXR fragment containing its ligand binding domain
  • creatine kinase assays e.g., assays based on the differentiation of pre-adipocytes
  • assays based on glucose uptake control in adipocytes e.g., glucose uptake control in adipocytes
  • immunological assays e.g., assays for examining binding of test agents to
  • subject includes mammals, especially humans. It also encompasses other non-human animals that are amenable for treatment with LXR agonists of the present invention.
  • a "variant" of a molecule such as an LXR receptor or an LXR agonist is meant to refer to a molecule substantially similar in structure and biological activity to either the entire molecule, or to a fragment thereof. Thus, provided that two molecules possess a similar activity, they are considered variants as that term is used herein even if the composition or secondary, tertiary, or quaternary structure of one of the molecules is not identical to that found in the other, or if the sequence of amino acid residues is not identical.
  • a “vector” is a composition for facilitating introduction, replication and/ or expression of a selected nucleic acid in a cell.
  • Vectors include, e.g., plasmids, cosmids, viruses, YACs, bacteria, poly-lysine, etc.
  • a "vector nucleic acid” is a nucleic acid molecule into which heterologous nucleic acid is optionally inserted which can then be introduced into an appropriate host cell.
  • Vectors preferably have one or more origins of replication, and one or more sites into which the recombinant DNA can be inserted.
  • Vectors often have convenient means by which cells with vectors can be selected from those without, e.g., they encode drug resistance genes.
  • Expression vectors are vectors that comprise elements that provide for or facilitate transcription of nucleic acids that are cloned into the vectors. Such elements can include, e.g., promoters and/or enhancers operably coupled to a nucleic acid of interest.
  • LXR agonists that are suitable for practicing methods of the present invention. They can be known agents that activate LXR receptor, e.g., GW3965 (see Examples below), or other commercially available compounds such as F3MethylAA (from Merck; see Menke et al., Endocrinology 143: 2548-58, 2002) and T0901317 (Tularik, California; see Examples below). They can also be novel LXR agonists to be screened for in accordance with the present invention. As detailed below, the LXR agonists suitable for the present invention can be polypeptides, peptides, small molecules, or other agents. The LXR agonists can be agonists for LXR of human as well as other animals.
  • LXR agonists have been described in the art.
  • small molecule LXR agonists include the well known oxysterols and related compounds (Janowski et al., Nature 383: 728-31, 1996); T0901317 and T0314407 (Schultz et al., Genes Dev 14: 2831-8, 2000); 24(S)-hydroxycholesterol, and 22(R)-hydroxycholesterol (Janowski et al, Nature 383: 728-731, 1996); and 24,25-epoxycholesterol (US Patent No. 6,316,503).
  • Exemplary polypeptide agonists of LXR have also been disclosed in the art, e.g., WO 02/077229.
  • LXR agonists are effective in activating both LXR ⁇ and LXR ⁇ (e.g., GW3965 as described in Collins et al., J Med Chem. 45: 1963-6, 2002).
  • Some LXR agonists activate LXR ⁇ and LXR ⁇ under different conditions.
  • 6-alpha-hydroxylated bile acids are agonists of LXR ⁇ , but also activate LXR ⁇ at higher concentrations (Song et al., Steroids 65: 423-7, 2000).
  • Some LXR agonists act exclusively on LXR ⁇ , while some others activate only LXR ⁇ .
  • LXR agonists can also be obtained from derivatives of known polypeptide agonists of the LXR receptor. They can be produced by a variety of art known techniques. For example, specific oligopeptides (e.g., 10-25 amino acid residues) spanning a known polypeptide agonist of LXR can be synthesized (e.g., chemically or recombinantly) and tested for their ability to activate an LXR receptor.
  • the LXR agonist fragments can be synthesized using standard techniques such as those described in Bodansky, M. Principles of Peptide Synthesis, Springer Verlag, Berlin (1993) and Grant, G. A (ed.). Synthetic Peptides: A User's Guide, W. H.
  • LXR agonists can be produced by digestion of native or recombinantly produced polypeptide agonists of LXR using a protease, e.g., trypsin, thermolysin, chymotrypsin, or pepsin.
  • a protease e.g., trypsin, thermolysin, chymotrypsin, or pepsin.
  • Computer analysis using commercially available software, e.g. Mac Vector, Omega, PCGene, Molecular Simulation, Inc. can be used to identify proteolytic cleavage sites.
  • polypeptide or peptide agonists for use in methods of the present invention are preferably isolated and substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the LXR agonists is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized.
  • the proteolytic or synthetic polypeptide agonists or their fragments can comprise as many amino acid residues as are necessary to activate LXR receptor activity, and can comprise at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or more amino acids in length.
  • LXR agonists can also be obtained by screening test agents (e.g., compound libraries) to identify novel LXR agonists that bind to and/or activate LXR receptor activities.
  • test agents e.g., compound libraries
  • novel LXR agonists e.g., a human LXR or LXR of other animals can be employed in a proper assay system.
  • Polynucleotide and amino acid sequences of the LXR receptors are known and described in the art. Their structures and functional organizations, including their ligand binding domains, have also been characterized.
  • the agonists can activate either LXR or LXR ⁇ .
  • some of the screen assays can employ an LXR polypeptide that comprises a fragment of an LXR molecule.
  • LXR polypeptide that comprises a fragment of an LXR molecule.
  • the two functional domains of the LXR receptor, the N-terminal DNA binding domain (DBD) and the C-terminal ligand- binding domain (LBD) mediate the transcriptional activation function of nuclear receptors.
  • An LXR polypeptide containing any of these domains can be used in screening for novel LXR agonists.
  • test agents can be screened for direct binding to an LXR polypeptide or a fragment thereof (e.g., its ligand binding domain).
  • potential LXR agonists can be examined for ability to activate LXR receptor pathway or stimulate other biological activities of the LXR receptor.
  • Either an in vitro assay system or a cell-based assay system can be used in the screening.
  • LXR ⁇ , LXR ⁇ , RXR, or PPAR LXR radioligand competition scintillation proximity assays described in, e.g., WO 01/41704, and the PPAR competition binding assays described in, e.g., Berger et al, J Biol Chem 274: 6718- 6725, 1999).
  • Test agents that can be screened for novel LXR agonists include polypeptides, beta-turn mimetics, polysaccharides, phospholipids, hormones, prostaglandins, steroids, aromatic compounds, heterocyclic compounds, benzodiazepines, oligomeric N- substituted glycines, oligocarbamates, polypeptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof.
  • Some test agents are synthetic molecules, and others natural molecules.
  • Test agents are obtained from a wide variety of sources including libraries of synthetic or natural compounds.
  • Combinatorial libraries can be produced for many types of compound that can be synthesized in a step-by-step fashion.
  • Large combinatorial libraries of compounds can be constructed by the encoded synthetic libraries (ESL) method described in WO 95/12608, WO 93/06121, WO 94/08051, WO 95/35503 and WO 95/30642.
  • Peptide libraries can also be generated by phage display methods (see, e.g., Devlin, WO 91/18980).
  • Libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts can be obtained from commercial sources or collected in the field.
  • Known pharmacological agents can be subject to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification to produce structural analogs.
  • Combinatorial libraries of peptides or other compounds can be fully randomized, with no sequence preferences or constants at any position.
  • the library can be biased, i.e., some positions within the sequence are either held constant, or are selected from a limited number of possibilities.
  • the nucleotides or amino acid residues are randomized within a defined class, for example, of hydrophobic amino acids, hydrophilic residues, sterically biased (either small or large) residues, towards the creation of cysteines, for cross-linking, prolines for SH-3 domains, serines, threonines, tyrosines or histidines for phosphorylation sites, or to purines.
  • test agents can be naturally occurring proteins or their fragments. Such test agents can be obtained from a natural source, e.g., a cell or tissue lysate. Libraries of polypeptide agents can also be prepared, e.g., from a cDNA library commercially available or generated with routine methods.
  • the test agents can also be peptides, e.g., peptides of from about 5 to about 30 amino acids, with from about 5 to about 20 amino acids being preferred, and from about 7 to about 15 being particularly preferred.
  • the peptides can be digests of naturally occurring proteins, random peptides, or "biased" random peptides. In some methods, the test agents are polypeptides or proteins.
  • test agents can also be nucleic acids.
  • Nucleic acid test agents can be naturally occurring nucleic acids, random nucleic acids, or "biased" random nucleic acids. For example, digests of prokaryotic or eukaryotic genomes can be similarly used as described above for proteins.
  • the test agents are small organic molecules (e.g., molecules with a molecular weight of not more than about 1,000).
  • high throughput assays are adapted and used to screen for such small molecules.
  • combinatorial libraries of small molecule test agents as described above can be readily employed to screen for small molecule modulators of an LXR receptor.
  • a number of assays are available for such screening, e.g., as described in Schultz (1998) Bioorg Med Chem Lett 8:2409-2414; Weller (1997) Mol Divers. 3:61-70; Fernandes (1998) Curr Opin Chem Biol 2:597-603; and Sittampalam (1997) Curr Opin Chem Biol 1 : 384-91.
  • LXR agonists can also be identified based on rational design. For example, Janowski et al. (Proc atl Acad Sci USA 96: 266-71, 1999) disclosed structural requirements of ligands for LXR ⁇ and LXR ⁇ . It was shown that position-specific monooxidation of the sterol side chain of oxysterol is requisite for LXR high-affinity binding and activation. Enhanced binding and activation can also be achieved through the use of 24- oxo ligands that act as hydrogen bond acceptors in the side chain. In addition, introduction of an oxygen on the sterol B-ring results in a ligand with LXR ⁇ -subtype selectivity.
  • Test agents to be screened with the claimed methods can also be generated based on structural studies of the LXR receptors, their fragments or analogs. Such structural studies allow the identification of test agents that are more likely to bind to the LXR receptor.
  • the three-dimensional structure of an LXR receptor can be studied in a number of ways, e.g., crystal structure and molecular modeling. Methods of studying protein structures using x-ray crystallography are well known in the literature. See Physical Bio-chemistry, Van Holde, K. E. (Prentice-Hall, New Jersey 1971), pp. 221-239, and Physical Chemistry with Applications to the Life Sciences, D. Eisenberg & D. C. Crothers (Benjamin Cummings, Menlo Park 1979).
  • binding of a test agent to an LXR or an LXR polypeptide containing its ligand binding domain is determined. Binding of test agents (e.g., polypeptides) to the LXR polypeptide can be assayed by a number of methods including, e.g., labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, functional assays (phosphorylation assays, etc.), and the like. See, e.g., U.S.
  • test agent can be identified by detecting a direct binding to the LXR polypeptide, e.g., co-immunoprecipitation with the LXR polypeptide by an antibody directed to the LXR polypeptide.
  • the test agent can also be identified by detecting a signal that indicates that the agent binds to the LXR polypeptide, e.g., fluorescence quenching.
  • Competition assays provide a suitable format for identifying test agents (e.g., peptides or small molecule compounds) that specifically bind to an LXR polypeptide.
  • test agents are screened in competition with a compound already known to bind to the LXR polypeptide.
  • the known binding compound can be a synthetic compound. It can also be an antibody, which specifically recognizes the LXR polypeptide, e.g., a monoclonal antibody directed against the LXR polypeptide. If the test agent inhibits binding of the compound known to bind the LXR polypeptide, then the test agent also binds the LXR polypeptide.
  • RIA solid phase direct or indirect radioimmunoassay
  • EIA solid phase direct or indirect enzyme immunoassay
  • sandwich competition assay see Stahli et al., Methods in Enzymology 9:242-253 (1983)
  • solid phase direct biotin-avidin EIA see Kirkland et al., J. Immunol. 137:3614-3619 (1986)
  • solid phase direct labeled assay solid phase direct labeled sandwich assay
  • solid phase direct labeled sandwich assay see Harlow and Lane, "Antibodies, A Laboratory Manual," Cold Spring Harbor Press (1988)
  • solid phase direct label RIA using 125 I label see Morel et al., Mol. Immunol.
  • Modulating agents identified by competition assay include agents binding to the same epitope as the reference compound and agents binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference compound for steric hindrance to occur. Usually, when a competing agent is present in excess, it will inhibit specific binding of a reference compound to a common target polypeptide by at least 50 or 75%.
  • the screening assays can be either in insoluble or soluble formats.
  • One example of the insoluble assays is to immobilize an LXR polypeptide or its fragments onto a solid phase matrix.
  • the solid phase matrix is then put in contact with test agents, for an interval sufficient to allow the test agents to bind. After washing away any unbound material from the solid phase matrix, the presence of the agent bound to the solid phase allows identification of the agent.
  • the methods can further include the step of eluting the bound agent from the solid phase matrix, thereby isolating the agent.
  • the test agents are bound to the solid matrix and the LXR polypeptide molecule is then added.
  • Soluble assays include some of the combinatory libraries screening methods described above. Under the soluble assay formats, neither the test agents nor the LXR polypeptide are bound to a solid support. Binding of an LXR polypeptide or fragment thereof to a test agent can be determined by, e.g., changes in fluorescence of either the LXR polypeptide or the test agents, or both. Fluorescence may be intrinsic or conferred by labeling either component with a fluorophor.
  • either the LXR polypeptide, the test agent, or a third molecule can be provided as labeled entities, i.e., covalently attached or linked to a detectable label or group, or cross-linkable group, to facilitate identification, detection and quantification of the polypeptide in a given situation.
  • detectable groups can comprise a detectable polypeptide group, e.g., an assayable enzyme or antibody epitope.
  • the detectable group can be selected from a variety of other detectable groups or labels, such as radiolabels (e.g., I, P, S ) or a chemiluminescent or fluorescent group.
  • the detectable group can be a substrate, cofactor, inhibitor or affinity ligand.
  • Binding of a test agent to LXR can also be tested indirectly with a cell-based assay.
  • a DNA-binding domain of the nonreceptor transcription factor GAL4 can be fused to the ligand-binding domain of LXR (e.g., LXR ⁇ ).
  • the resultant construct is introduced into a host cell (e.g., the 293 cells) together with a reporter construct (e.g., a UAS-containing luciferase reporter construct).
  • reporter construct e.g., a UAS-containing luciferase reporter construct.
  • reporter polypeptide activity e.g., luciferase activity
  • Effects of individual test agents on the reporter polypeptide activity are evaluated relative to a control (i.e., when no test compound is present).
  • the cell-free ligand sensing assay can also be employed to identify novel LXR agonists. It can be performed as described in the art, e.g., Collins et al., J Med Chem. 45: 1963-6, 2002; and Spencer et al., J Med. Chem. 44: 886-97, 2001.
  • This assay measures the ligand-dependent recruitment of a peptide from the steroid receptor coactivator 1 (SRC1) to the nuclear receptor.
  • SRC1 steroid receptor coactivator 1
  • LXR agonists for use in the methods of the present invention can also be examined for ability to activate other bioactivities or cellular activities of the LXR receptor.
  • Test agents which activate LXR receptor can be identified by monitoring their effects on a number of LXR cellular activities.
  • LXR cellular activities include any activity mediated by activated LXR receptor (e.g., transcriptional regulation of a target gene).
  • LXR trans-activate expression of a number of target genes e.g., ABCA1
  • a target genes e.g., ABCA1
  • modulate the production of muscle-specific enzymes e.g., creatine kinase
  • modulate glucose uptake by cells and stimulate myoblast cell proliferation.
  • the degree to which a test agent activates an LXR receptor can be identified by testing for the ability of the agent to enhance such LXR activities.
  • a novel LXR agonist can be identified by identifying a test agent that enhances expression of an LXR target gene (e.g., ABCA1, ABCG1, SREBP1, or the cholesterol 7- hydroxylase gene).
  • LXR target gene e.g., ABCA1, ABCG1, SREBP1, or the cholesterol 7- hydroxylase gene.
  • LXR agonists can also be identified by examining other cellular activities stimulated by the LXR pathway. For example, LXR agonists modulate the protein level and hence activity of a muscle-specific enzyme, creatine kinase. Therefore, LXR agonists can be screened by examining test agents for ability to modulate creatine kinase activity, e.g., as described in Somjen et al., J Steroid Biochem Mol Biol 62: 401-8, 1997. The assay can be performed in a cell line, e.g., the mouse skeletal myoblast cell line or a primary chick myoblast cell line. Effects of test compounds on creatine kinase activity in the cultured cells can be measured in the cell lysates using a commercially available kit (available by Sigma, St Louis, Mo.).
  • Modulation of other cellular bioactivities of the LXR receptor can also be detected using methods well known and routinely practiced in the art.
  • the test agent can be assayed for their activities in increasing cholesterol efflux from cells such as macrophages (Menke et al., Endocrinology 143: 2548-58, 2002; and Sparrow et al., J Biol. Chem. 277: 10021-7, 2002).
  • assays include ligand-dependent transcription assays (Schmidt et al., Mol Cell Endocrinol 155: 51-60, 1999), methods for measuring the ability of LXR agonists to interfere with the differentiation process of pre-adipocytes (fibroblasts) to adipocytes (Plaas et al., Biosci Rep 1: 207-16, 1981; Hiragun et al., J Cell Physiol 134: 124- 30, 1988; and Liao et al., J Biol Chem 270: 12123-32, 1995), or the ability to stimulate myoblast cell proliferation (Konishi et al, Biochemistry 28: 8872-7, 1989; and Austin et al., J Neurol Sci 101: 193-7, 1991). As a control, all these assays can include measurements before and after the test agent is added to the assay system.
  • the present invention provides methods for modulating expression of genes involved in glucose transport (e.g., glut4) and gluconeogenesis (e.g., PGC-1 or PEPCK). These methods of the invention can be used either in vitro or in vivo to increase insulin sensitivity and/or glucose uptake by a cell, as well as reducing glucose output by the liver cells. The methods also find application in treating a disease characterized by insufficient glut4 expression, insulin dysfunction (e.g., resistance, inactivity or deficiency) and/or insufficient glucose transport into cells. Such diseases include, but are not limited to diabetes, hyperglycemia and obesity.
  • Modulation of glut4 expression is also useful for preventing or modulating the development of such diseases or disorders in a subject suspected of being, or known to be, prone to such diseases or disorders.
  • the LXR agonists to be used in these applications can be any of the known LXR agonists that have been described in the art.
  • the therapeutic methods comprise screening test agents to identify novel LXR agonists as described above, and administering such novel agonists to enhance glut4 expression in cells or to treat the above noted diseases in a subject.
  • Methods of the present invention can be used to modulate of expression of a number of genes that are involved in glucose metabolism.
  • the invention provides methods for enhancing glut4 expression in fat cells or muscle cells such as white adipose cells and smooth muscle cells.
  • liver expression of gluconeogenesis- related genes can be inhibited or reduced in accordance with methods of the invention.
  • genes include PGC-1, PEPCK, glucose-6-phosphatase, and glucokinase.
  • Cells suitable for modulation include isolated cells maintained in culture, as well as cells within their natural context in vivo in a subject, e.g., in the liver, fat tissue or muscle tissue such as pectoralis, triceps, gastrocnemius, quadriceps, and iliocostal muscles of a mammal.
  • a cell can be contacted with any a number of the known LXR agonists or novel LXR agonists identified in accordance with the present invention.
  • an LXR agonist is introduced directly to a subject (e.g., a human or a non-human subject).
  • a polynucleotide encoding a polypeptide agonist of an LXR receptor is introduced by retroviral or other means (as detailed below).
  • an LXR agonist specific for the LXR ⁇ receptor is used.
  • an LXR ⁇ receptor-specific agonist is employed.
  • agonists that can activate both LXR ⁇ and LXR ⁇ are administered to cells to modulate the gene expression.
  • the cell is first determined to have low expression level of the relevant gene (e.g., glut4 level in an adipose cell) as compared to normal level ("baseline level,” or "a desired level") of the same cell type.
  • expression levels of the genes are measured before and/or after treatment with the LXR agonist in order to confirm that the treatment results in modulated expression level of the genes.
  • the in vivo effect can be monitored by taking a tissue sample from the subject and analyzing expression levels of the genes to be modulated, e.g., glut4 in adipose tissue or PGC-1 in liver cells.
  • the tissue or cell samples can be obtained by following the well-established and routinely practiced medical procedures.
  • Animal adipose tissue sample (e.g., needle biopsy from subcutaneous adipose tissue) can be easily obtained as described in, e.g., Martinsson et al., J Med Lab Technol, 24: 52-3, 1967; Novak et al, Exp Cell Res 73: 335-44, 1972; and Taskinen et al, Clin Chim Acta 104: 107- 17, 1980.
  • Percutaneous adipose tissue biopsy can be obtained by mini-liposuction method as described in, e.g., Bastard et al., J Parenter Enteral Nutr 18: 466-8, 1994.
  • a small liver tissue sample from a subject can be obtained by the well established liver biopsy methods as described in e.g., Oxender et al., J Dairy Sci 54: 286-8, 1971; Spiezia et al., Eur J Ultrasound 15: 127-31, 2002; and Rinella et al., Liver Transpl. 8: 1123- 5, 2002.
  • transgenic animals with integrated human genes (e.g., glut4 or PGC-1) and LXR-encoding sequences can be used to assay induction of glut4 expression in vivo.
  • Transgenic animals e.g., transgenic mice harboring the human sequences can be generated according to methods well known in the art. For example, techniques routinely used to create and screen for transgenic animals have been described in, e.g., see Bijvoet (1998) Hum. Mol. Genet. 7:53-62; Moreadith (1997) J. Mol.
  • the present invention also encompasses therapeutic methods for treating or ameliorating diabetes mellitus and related disorders such as obesity or hyperglycemia.
  • diabetes mellitus especially type II diabetes mellitus is well documented (J Clin Endocrinol Metab 77: 25-6, 1993).
  • Glut4 is primarily expressed in adipose and muscle tissues. It was suggested that a reduction in glut4 expression in slow fibers reduces the insulin-sensitive Glut4 pool in type II diabetes and thus contributes to skeletal muscle insulin resistance (Gaster et al., Diabetes 50: 1324-9, 2001).
  • Glut4 Muscle-specific inactivation of Glut4 caused glucose toxicity and the development of diabetes in mice (Kim et al., J Clin Invest 108: 153-60, 2001).
  • a compound which improves peripheral insulin resistance in type II diabetic subjects and animal models apparently exerts beneficial effects by increasing glut4 expression in adipose tissue (Furuta et al., Diabetes Res Clin Pract 56: 159-71, 2002).
  • administration of LXR agonists to a subject suffering from diabetes e.g., type II diabetes
  • therapeutical effects are monitored by measuring circulating glucose level in the subject before and/or after administering an LXR agonist.
  • Glucose level in the subject can be measured with methods well known in the art. For example, blood glucose levels can be measured very simply and quickly with many commercially available blood glucose testing kits.
  • the present inventors observed that modulation of expression of glucose metabolism-related genes can be achieved after application of an LXR agonist for a very short period of time, e.g., in 3 days.
  • an LXR agonist is typically administered to a subject for a continued period of time, e.g., at least 10 days, 14 days, 30 days, 60 days, 90 days, or longer.
  • the LXR agonists of the present invention can be directly administered under sterile conditions to the subject to be treated.
  • the modulators can be administered alone or as the active ingredient of a pharmaceutical composition.
  • Therapeutic composition of the present invention can be combined with or used in association with other therapeutic agents.
  • a subject may be treated with an LXR agonist along with other conventional anti-diabetes drugs.
  • Examples of such known anti-diabetes drugs include Actos (pioglitizone, Takeda, Eli Lilly ), Avandia (rosiglitazone, Smithkline Beacham), Amaryl (glimepiride, Aventis), Glipizide Sulfonlyurea (Generic) or Glucotrol (Pfizer), Glucophage (metformin, Bristol Meyers Squibb), Glucovance (glyburide/metformin, Bristol Meyers Squibb), Glucotrol XL (glipizide extended release, Pfizer), Glyburide (Micronase; Upjohn, Glynase; Upjohn, Diabeta; Aventis), Glyset (miglitol, Pharmacia & Upjohn), Metaglip (glipizide + metformin; fixed combination tablet), Prandin (repaglinide, NOVO), Precose (acarbose, Bayer), Rezulin (troglitazone, Park
  • compositions of the present invention typically comprise at least one active ingredient together with one or more acceptable carriers thereof.
  • Pharmaceutically carriers enhance or stabilize the composition, or to facilitate preparation of the composition.
  • Pharmaceutically acceptable carriers are determined in part by the particular composition being administered (e.g., nucleic acid, protein, or modulatory compounds), as well as by the particular method used to administer the composition. They should also be both pharmaceutically and physiologically acceptable in the sense of being compatible with the other ingredients and not injurious to the subject.
  • This carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral, sublingual, rectal, nasal, or parenteral.
  • suitable pharmaceutically acceptable carriers include syrup, water, isotonic saline solution, 5% dextrose in water or buffered sodium or ammonium acetate solution, oils, glycerin, alcohols, flavoring agents, preservatives, coloring agents starches, sugars, diluents, granulating agents, lubricants, and binders, among others.
  • the LXR agonist can also be complexed with carrier proteins such as ovalbumin or serum albumin prior to their administration in order to enhance stability.
  • compositions can be prepared in various forms, such as granules, tablets, pills, suppositories, capsules, suspensions, salves, lotions and the like.
  • concentration of therapeutically active compound in the formulation may vary from about 0.1-100% by weight.
  • Therapeutic formulations are prepared by any methods well known in the art of pharmacy.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • suitable routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • parenteral administration the LXR agonists of the present invention maybe formulated in a variety of ways.
  • Aqueous solutions of the modulators may be encapsulated in polymeric beads, nanoparticles or other injectable depot formulations known to those of skill in the art. Additionally, the compounds of the present invention may also be administered encapsulated in liposomes.
  • compositions may be present both in the aqueous layer and in the lipidic layer, or in what is generally termed a liposomic suspension.
  • the hydrophobic layer generally but not exclusively, comprises phospholipids such as lecithin and sphingomyelin, steroids such as cholesterol, more or less ionic surfactants such a diacetylphosphate, stearylamine, or phosphatidic acid, and/or other materials of a hydrophobic nature.
  • compositions may be supplemented by other active pharmaceutical ingredients, where desired.
  • Optional antibacterial, antiseptic, and antioxidant agents may also be present in the compositions where they will perform their ordinary functions.
  • the LXR agonists are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a sustained release material such as glyceryl monostearate or glyceryl distearate, alone or with a wax can be included in the compositions.
  • Biodegradable, biocompatible polymers can also be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • Subjects suffering from diabetes or related disorders are typically treated with pharmaceutical compositions of the present invention for a continued period of time (e.g., at least 10 days, 14 days, 30 days, 60 days, 90 days, or longer).
  • the pharmaceutical compositions comprise a pharmaceutically effective amount or prophylactically effective amount of an LXR agonist.
  • therapeutically effective amount is intended to mean that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, a system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
  • prophylactically effective amount is intended to mean that amount of a pharmaceutical drug that will prevent or reduce the risk of occurrence of the biological or medical event that is sought to be prevented in a tissue, a system, animal or human.
  • a suitable therapeutic dose can be determined by any of the well-known methods such as clinical studies on mammalian species to determine maximum tolerable dose and on normal human subjects to determine safe dosage.
  • the dosage amount of an LXR ligand that a subject receives can be selected so as to achieve the desired up-regulation of glut4 expression; the dosage a subject receives may also be titrated over time in order to reach a target Glut4 level.
  • Toxicity and therapeutic efficacy of LXR agonists can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 / ED 50 .
  • LXR agonists that exhibit large therapeutic indices are preferred. While LXR agonists that exhibit toxic side.effects may be used, care should be taken to design a delivery system that targets such LXR agonists to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosages for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the ICso (i.e., the concentration of the test LXR agonists which achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • ICso i.e., the concentration of the test LXR agonists which achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • the preferred dosage of an LXR agonist usually lies within the range of from about 0.001 to about 1000 mg, more usually from about 0.01 to about 500 mg per day.
  • the preferred dosage and mode of administration of an LXR agonist can vary for different subjects, depending upon factors that can be individually reviewed by the treating physician, such as the condition or conditions to be treated, the choice of composition to be administered, including the particular LXR agonist, the age, weight, and response of the individual subject, the severity of the subject's symptoms, and the chosen route of administration.
  • the quantity of an LXR agonist administered is the smallest dosage that effectively and reliably prevents or minimizes the conditions of the subjects.
  • a first LXR agonist is used in combination with a second LXR agonist or a known anti-diabetes drug in order to achieve therapeutic effects that cannot be achieved when just one LXR agonist is used individually.
  • polynucleotides encoding LXR agonists of the present invention are transfected into cells for therapeutic purposes in vitro and in vivo. These polynucleotides can be inserted into any of a number of well-known vectors for the transfection of target cells and organisms as described below. The polynucleotides are transfected into cells, ex vivo or in vivo, through the interaction of the vector and the target cell. The compositions are administered to a subject in an amount sufficient to elicit a therapeutic response in the subject.
  • a polynucleotide encoding the LXR receptor can be transfected into cells or administered to a subject for therapeutic purposes.
  • the subject can be further administered an LXR agonist (e.g., a small molecule LXR agonist) as described above.
  • LXR agonist e.g., a small molecule LXR agonist
  • Expression of the exogenous LXR receptors and administration of the LXR agonist could stimulate LXR mediated pathway, including regulation of glucose metabolism.
  • Non- viral vector delivery systems include DNA plasmids, naked nucleic acid, and nucleic acid complexed with a delivery vehicle such as a liposome.
  • Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell.
  • Methods of non- viral delivery of nucleic acids include lipofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipidmucleic acid conjugates, naked DNA, artificial virions, and agent- enhanced uptake of DNA.
  • Lipofection is described in, e.g., US Pat. No. 5,049,386, US Pat. No. 4,946,787; and US Pat. No. 4,897,355, and lipofection reagents are sold commercially (e.g., TransfectamTM and LipofectinTM).
  • Cationic and neutral lipids that are suitable for efficient receptor- recognition lipofection of polynucleotides include those described in WO 91/17424 and WO 91/16024. Delivery can be directed to cells (ex vivo administration) or target tissues (in vivo administration).
  • lipid:nucleic acid complexes including targeted liposomes such as immunolipid complexes
  • the preparation of lipid:nucleic acid complexes, including targeted liposomes such as immunolipid complexes, is well known to one of skill in the art (see, e.g., Crystal, Science 270:404-410 (1995); Blaese et al, Cancer Gene Ther. 2:291-297 (1995); Behr et al, Bioconjugate Chem. 5:382-389 (1994); Remy et al, Bioconjugate Chem. 5:647-654 (1994); Gao et al, Gene Therapy 2:710-722 (1995); Ahmad et al, Cancer Res. 52:4817-4820 (1992); U.S. Pat. Nos. 4,186,183, 4,217,344, 4,235,871, 4,261,975, 4,485,054, 4,501,728, 4,774,085, 4,837,028, and 4,946,787).
  • Gene therapy vectors can be delivered in vivo by administration to an individual subject, typically by systemic administration (e.g., intravenous, intraperitoneal, intramuscular, subdermal, or intracranial infusion) or topical application, as described below.
  • vectors can be delivered to cells ex vivo, such as cells explanted from an individual subject (e.g., lymphocytes, bone marrow aspirates, tissue biopsy) or universal donor hematopoietic stem cells, followed by reimplantation of the cells into a subject, usually after selection for cells which have incorporated the vector.
  • Ex vivo cell transfection for diagnostics, research, or for gene therapy is well known to those of skill in the art.
  • cells are isolated from the subject organism, transfected with a nucleic acid (gene or cDNA), and re-infused back into the subject organism (e.g., subject).
  • a nucleic acid gene or cDNA
  • Various cell types suitable for ex vivo transfection are well known to those of skill in the art (see, e.g., Freshney et al, Culture of Animal Cells, A Manual of Basic Technique (3rd ed. 1994)) and the references cited therein for a discussion of how to isolate and culture cells from subjects).
  • Vectors e.g., retro viruses, adenoviruses, liposomes, etc.
  • therapeutic nucleic acids can be also administered directly to the organism for transduction of cells in vivo.
  • naked DNA can be administered.
  • Administration is by any of the routes normally used for introducing a molecule into ultimate contact with blood or tissue cells. Suitable methods of administering such nucleic acids are available and well known to those of skill in the art, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route.
  • RNA or DNA viral based systems for the delivery of nucleic acids take advantage of highly evolved processes for targeting a virus to specific cells in the body and trafficking the viral payload to the nucleus.
  • Viral vectors can be administered directly to subjects (in vivo) or they can be used to treat cells in vitro and the modified cells are administered to subjects (ex vivo).
  • Conventional viral based systems for the delivery of nucleic acids could include retroviral, lentivirus, adenoviral, adeno-associated and herpes simplex virus vectors for gene transfer.
  • Viral vectors are currently the most efficient and versatile method of gene transfer in target cells and tissues. Integration in the host genome is possible with the retrovirus, lentivirus, and adeno-associated virus gene transfer methods, often resulting in long term expression of the inserted transgene. Additionally, high transduction efficiencies have been observed in many different cell types and target tissues.
  • Lentiviral vectors are retroviral vector that are able to transduce or infect non-dividing cells and typically produce high viral titers. Selection of a retroviral gene transfer system would therefore depend on the target tissue. Retroviral vectors are comprised of cw-acting long terminal repeats with packaging capacity for up to 6-10 kb of foreign sequence. The minimum c ⁇ -acting LTRs are sufficient for replication and packaging of the vectors, which are then used to integrate the therapeutic gene into the target cell to provide permanent transgene expression.
  • Widely used retroviral vectors include those based upon murine leukemia virus (MuLV), gibbon ape leukemia virus (GaLV), simian immunodeficiency virus (SIN), human immunodeficiency virus (HIN), and combinations thereof (see, e.g., Buchscher et al, J. Virol. 66:2731-2739 (1992); Johann et /., J. Virol. 66:1635-1640 (1992); Sommerfelt et ⁇ /., Virol. 176:58-59 (1990); Wilson et al., J. Virol 63:2374-2378 (1989); Miller et al, J. Virol. 65:2220-2224 (1991); PCT/US94/05700).
  • MiLV murine leukemia virus
  • GaLV gibbon ape leukemia virus
  • SIN simian immunodeficiency virus
  • HIN human immunodeficiency virus
  • pLAS ⁇ and MFG-S are examples are retroviral vectors that have been used in clinical trials (Dunbar et al, Blood 85:3048-305 (1995); Kohn et al, Nat. Med. 1:1017-102 (1995); Malech et al, Proc. Natl. Acad. Sci. U.S.A. 94:22 12133-12138 (1997)).
  • PA317/pLAS ⁇ was the first therapeutic vector used in a gene therapy trial.
  • a viral vector is typically modified to have specificity for a given cell type by expressing a ligand as a fusion protein with a viral coat protein on the viruses outer surface.
  • the ligand is chosen to have affinity for a receptor known to be present on the cell type of interest.
  • Han et al. Proc. Natl. Acad. Sci. U.S.A. 92:9747-9751 (1995), reported that Moloney murine leukemia virus can be modified to express human heregulin fused to gp70, and the recombinant virus infects certain human breast cancer cells expressing human epidermal growth factor receptor.
  • filamentous phage can be engineered to display antibody fragments (e.g., FAB or Fv) having specific binding affinity for virtually any chosen cellular receptor.
  • FAB fragment-binding protein
  • Fv antibody fragment-binding protein
  • FIG. 1 A shows that LXR ligand GW3965 induces glucokinase expression and represses genes involved in gluconeogenesis in liver.
  • the LXR ligand also induces GLUT4 expression in white adipose tissue (Fig. IB).
  • Figure 2 A shows that LXR ligands regulate ABCAl, but do not alter GLUT4 or PGC-1 expression in skeletal muscle. Effects of LXR ligands on expression of adipocyte signaling molecules are shown in Fig. 2B.
  • This Example shows that LXR agonists regulate GLUT4 and PGC-1 expressions in a cell autonomous manner.
  • Cells were treated with vehicle or 1 ⁇ M T1317, 1 ⁇ M GW3965, 2 ⁇ M 22(R)-hydroxycholesterol or 50 nM LG268 for 24 hours as indicated.
  • mRNA expression was determined by real time quantitative PCR assays.
  • the results indicate that LXR ligands repress PGC-1 expression in primary human hepatocytes (Fig. 5 A) and induce GLUT4 expression in 3T3-L1 adipocytes (Fig. 5B).
  • Fig. 6A shows modulation of Glut4 expression in macrophages by synthetic and oxysterol LXR ligands. Regulation of Glut4 expression by the LXR ligands is abolished in cells from LXR null mice.
  • Fig. 7 shows a conserved DR-4 hormone response element in the mouse Glut4 promoter (SEQ ID NO: 1) and human Glut4 promoter (SEQ ID NO: 2).
  • electromobility shift assays were performed using in vitro translated proteins and radiolabeled mGLUT4 oligonucleotide (Fig. 8A). The results indicate a functional LXR binding site in the GLUT4 promoter.
  • mice were fed a high fat diet for three months (Clinton/Cybulsky rodent diet; 40% kcal from fat, devoid of cholesterol) to induce obesity. After 3 months, mice were treated for one week with vehicle or 20 mg/kg/day GW3965. Glucose tolerance tests were performed by intraperitoneal injection of glucose (2 g/kg body weight) after 8 hours of fasting (Fig. 10A). In addition, effect of GW3965 on glucose tolerance in lean C57B1/6 mice was also examined (Fig. 10B). The lean C57B1/6 mice maintained on normal chow diet and similarly treated as indicated above. The results indicate that LXR ligands improve glucose tolerance and insulin resistance in diet-induced obesity.
  • mice Svnergistic Effects Between LXR Agonists and Anti-Diabetic Drugs
  • This Example describes treatment of mice with a combination of an LXR agonist and a known anti-diabetic drug.
  • Obese, insulin-resistant ob/ob mice were treated with an LXR ligand, GW3965, at 20 mg/kg/day and/or metformin at 300 mg/kg/day for 3 months. Fasting plasma glucose levels were then determined. The results indicate that mice treated with the combination of LXR ligand and metformin had circulating glucose level that is significantly lower than that in mice treated with either compound alone. As shown in Fig.
  • mice treated with the LXR agonist or metformin alone did not show significant difference from that of control (mice treated with vehicle).
  • mice were treated with a combination of GW3965 and metformin their plasma glucose level was significantly reduced as compared to that in the control mice.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Genetics & Genomics (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

La présente invention concerne de nouvelles méthodes de modulation de l'expression du glut4 et d'autres gènes impliqués dans le métabolisme du glucose, ainsi que des méthodes de traitement du diabète et d'autres maladies associées. Ces méthodes consistent à administrer à des cellules chez un sujet une dose utile d'un agoniste LXR pour moduler l'expression de ces gènes qui jouent un rôle important dans l'assimilation du glucose ou la gluconéogenèse. Cette modulation va augmenter l'assimilation du glucose par les cellules chez un sujet et/ou réduire la production de glucose dans le foie, et, par conséquent, atténuer les symptômes associés au diabète de type II.
PCT/US2003/040906 2002-12-23 2003-12-22 Nouvelle utilisation d'agonistes des recepteurs hepatiques x WO2004058175A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003301216A AU2003301216A1 (en) 2002-12-23 2003-12-22 Novel use of liver x receptor agonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US43611202P 2002-12-23 2002-12-23
US60/436,112 2002-12-23

Publications (2)

Publication Number Publication Date
WO2004058175A2 true WO2004058175A2 (fr) 2004-07-15
WO2004058175A3 WO2004058175A3 (fr) 2004-09-10

Family

ID=32682340

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/040906 WO2004058175A2 (fr) 2002-12-23 2003-12-22 Nouvelle utilisation d'agonistes des recepteurs hepatiques x

Country Status (3)

Country Link
US (1) US20050036992A1 (fr)
AU (1) AU2003301216A1 (fr)
WO (1) WO2004058175A2 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6924311B2 (en) * 2001-10-17 2005-08-02 X-Ceptor Therapeutics, Inc. Methods for affecting various diseases utilizing LXR compounds
WO2006112703A2 (fr) * 2005-04-20 2006-10-26 Academisch Medisch Centrum Bij De Universiteit Van Amsterdam Moyens et procedes permettant de contrebalancer l'accumulation d'acide gras
WO2007065444A1 (fr) * 2005-12-09 2007-06-14 Saibot Aps Méthode pour évaluer le potentiel d'un composé en vue de son utilisation en thérapie
JP2008540490A (ja) * 2005-05-10 2008-11-20 ラボラトワール フルニエ エス・アー 肝臓x受容体アゴニストの新規な使用
US8076376B2 (en) 2005-07-22 2011-12-13 Powers Jay P Aniline sulfonamide derivatives and their uses

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003223340A1 (en) * 2002-03-27 2003-10-13 Smithkline Beecham Corporation Certain pharmaceutically useful substituted aminoalkyl heterocycles
WO2004043939A1 (fr) * 2002-03-27 2004-05-27 Smithkline Beecham Corporation Composes amide et procedes d'utilisation de ceux-ci
WO2003082205A2 (fr) * 2002-03-27 2003-10-09 Smithkline Beecham Corporation Composes et procedes
US7560586B2 (en) * 2002-03-27 2009-07-14 Smithkline Beecham Corporation Acid and ester compounds and methods of using the same
CA2553443C (fr) * 2004-02-11 2012-03-27 Irm Llc Composes et compositions convenant comme modulateurs des lxr
US20070191371A1 (en) * 2006-02-14 2007-08-16 Kalypsys, Inc. Heterocyclic modulators of ppar
US20080070883A1 (en) * 2006-09-19 2008-03-20 Wyeth Use of LXR modulators for the prevention and treatment of skin aging
AU2007297721A1 (en) * 2006-09-19 2008-03-27 Wyeth Use of LXR agonists for the treatment of osteoarthritis
WO2009102789A2 (fr) * 2008-02-15 2009-08-20 Wyeth Utilisation d'agonistes de rxr pour le traitement de l'arthrose
WO2016057454A1 (fr) 2014-10-06 2016-04-14 The Johns Hopkins University Ciblage de récepteurs nucléaires de foie comme traitement pour la maladie de wilson
US20230132366A9 (en) 2018-11-26 2023-04-27 Denali Therapeutics Inc. Methods for treating dysregulated lipid metabolism

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6316503B1 (en) * 1999-03-15 2001-11-13 Tularik Inc. LXR modulators
US20020048572A1 (en) * 2000-05-03 2002-04-25 Bei Shan Treatment of hypertriglyceridemia and other conditions using LXR modulators
US20030073614A1 (en) * 2001-10-17 2003-04-17 Schulman Ira G. Methods for affecting various diseases utilizing LXR compounds
GB2381866A (en) * 2001-11-12 2003-05-14 Karobio Ab Assays for liver X receptor (LXR) modulators

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6316503B1 (en) * 1999-03-15 2001-11-13 Tularik Inc. LXR modulators
US20020048572A1 (en) * 2000-05-03 2002-04-25 Bei Shan Treatment of hypertriglyceridemia and other conditions using LXR modulators
US20030073614A1 (en) * 2001-10-17 2003-04-17 Schulman Ira G. Methods for affecting various diseases utilizing LXR compounds
GB2381866A (en) * 2001-11-12 2003-05-14 Karobio Ab Assays for liver X receptor (LXR) modulators

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6924311B2 (en) * 2001-10-17 2005-08-02 X-Ceptor Therapeutics, Inc. Methods for affecting various diseases utilizing LXR compounds
WO2006112703A2 (fr) * 2005-04-20 2006-10-26 Academisch Medisch Centrum Bij De Universiteit Van Amsterdam Moyens et procedes permettant de contrebalancer l'accumulation d'acide gras
WO2006112703A3 (fr) * 2005-04-20 2007-05-18 Amc Amsterdam Moyens et procedes permettant de contrebalancer l'accumulation d'acide gras
JP2008540490A (ja) * 2005-05-10 2008-11-20 ラボラトワール フルニエ エス・アー 肝臓x受容体アゴニストの新規な使用
US8076376B2 (en) 2005-07-22 2011-12-13 Powers Jay P Aniline sulfonamide derivatives and their uses
WO2007065444A1 (fr) * 2005-12-09 2007-06-14 Saibot Aps Méthode pour évaluer le potentiel d'un composé en vue de son utilisation en thérapie

Also Published As

Publication number Publication date
AU2003301216A8 (en) 2004-07-22
WO2004058175A3 (fr) 2004-09-10
US20050036992A1 (en) 2005-02-17
AU2003301216A1 (en) 2004-07-22

Similar Documents

Publication Publication Date Title
US20050036992A1 (en) Novel use of liver X receptor agonists
US20050037390A1 (en) Methods and compositions for modulating erythropoietin expression
Wang et al. C/EBPα arrests cell proliferation through direct inhibition of Cdk2 and Cdk4
US20080032961A1 (en) Therapy employing LXRalpha modolators
Kasembeli et al. TTI-101: A competitive inhibitor of STAT3 that spares oxidative phosphorylation and reverses mechanical allodynia in mouse models of neuropathic pain
US20030228607A1 (en) Screening method and modulators having an improved therapeutic profile
US20030203419A1 (en) Methods for screening and therapeutic applications of kinesin modulators
KR20010082559A (ko) 사람 Upf1p, 진핵세포 유리 인자 1 및 진핵세포 유리인자 3을 포함하는 감시 복합체가 연루된, 해독 종결 효율및 이상 mRNA 분해를 조절하는 방법
AU2005274377B2 (en) Method of screening for a carnitine transporter agonist or antagonist and its uses
JPH1192401A (ja) ヒト血清グルココルチコイドにより調節されるキナーゼ、慢性腎不全に対する標的
US20070231835A1 (en) Proteomic Screening for Redox State Dependent Protein-Protein Interactions
US7193053B2 (en) Hypoxia-inducible factor 1alpha variants and methods of use
TW201040282A (en) Utilizing shield effect for dopamine detection and reagent development
Shen et al. PLIN5 Suppresses Lipotoxicity and Ferroptosis in Cardiomyocyte via Modulating PIR/NF-κB Axis
US20040106148A1 (en) Polypeptides
EP1705256A1 (fr) Procédé de traitement de résistance à l'insuline et d'états pathologiques caractérisés par une résistance à l'insuline
JP2006525244A (ja) 糖尿病の治療標的としてのインスリン誘導性遺伝子
EP1438588B1 (fr) Methode d'identification d'agents moderateurs de la differenciation de pre-adipocytes
US20070054328A1 (en) Methods
US20050048585A1 (en) Methods and compositions for modulating NF-AT transcription factor
WO2004087888A2 (fr) Signalisation de neurotransmetteurs permettant la regulation de la duree de vie chez c. elegans
JP2005539208A (ja) グルコース産生を変化させるための組成物および方法
MXPA06006567A (en) Interaction of nmda receptor with the protein tyrosine phosphatase step in psychotic disorders

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP